Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Eur Urol Oncol ; 5(2): 164-175, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34774481

RESUMEN

BACKGROUND: No curative therapy is currently available for metastatic prostate cancer (PCa). The diverse mechanisms of progression include fibroblast growth factor (FGF) axis activation. OBJECTIVE: To investigate the molecular and clinical implications of fibroblast growth factor receptor 1 (FGFR1) and its isoforms (α/ß) in the pathogenesis of PCa bone metastases. DESIGN, SETTING, AND PARTICIPANTS: In silico, in vitro, and in vivo preclinical approaches were used. RNA-sequencing and immunohistochemical (IHC) studies in human samples were conducted. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: In mice, bone metastases (chi-square/Fisher's test) and survival (Mantel-Cox) were assessed. In human samples, FGFR1 and ladinin 1 (LAD1) analysis associated with PCa progression were evaluated (IHC studies, Fisher's test). RESULTS AND LIMITATIONS: FGFR1 isoform expression varied among PCa subtypes. Intracardiac injection of mice with FGFR1-expressing PC3 cells reduced mouse survival (α, p < 0.0001; ß, p = 0.032) and increased the incidence of bone metastases (α, p < 0.0001; ß, p = 0.02). Accordingly, IHC studies of human castration-resistant PCa (CRPC) bone metastases revealed significant enrichment of FGFR1 expression compared with treatment-naïve, nonmetastatic primary tumors (p = 0.0007). Expression of anchoring filament protein LAD1 increased in FGFR1-expressing PC3 cells and was enriched in human CRPC bone metastases (p = 0.005). CONCLUSIONS: FGFR1 expression induces bone metastases experimentally and is significantly enriched in human CRPC bone metastases, supporting its prometastatic effect in PCa. LAD1 expression, found in the prometastatic PCa cells expressing FGFR1, was also enriched in CRPC bone metastases. Our studies support and provide a roadmap for the development of FGFR blockade for advanced PCa. PATIENT SUMMARY: We studied the role of fibroblast growth factor receptor 1 (FGFR1) in prostate cancer (PCa) progression. We found that PCa cells with high FGFR1 expression increase metastases and that FGFR1 expression is increased in human PCa bone metastases, and identified genes that could participate in the metastases induced by FGFR1. These studies will help pinpoint PCa patients who use fibroblast growth factor to progress and will benefit by the inhibition of this pathway.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Próstata Resistentes a la Castración , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Factores de Crecimiento de Fibroblastos , Humanos , Masculino , Ratones , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
2.
Int J Cancer ; 148(8): 1928-1937, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33152115

RESUMEN

Survival of patients with relapsed/refractory osteosarcoma has not improved in the last 30 years. Several immunotherapeutic approaches have shown benefit in murine osteosarcoma models, including the anti-programmed death-1 (anti-PD-1) and anti-cytotoxic T-lymphocyte antigen-4 (anti-CTLA-4) immune checkpoint inhibitors. Treatment with the T-cell growth factor interleukin-2 (IL-2) has shown some clinical benefit but has limitations due to poor tolerability. Therefore, we evaluated the efficacy of bempegaldesleukin (BEMPEG; NKTR-214), a first-in-class CD122-preferential IL-2 pathway agonist, alone and in combination with anti-PD-1 or anti-CTLA-4 immune checkpoint inhibitors in metastatic and orthotopic murine models of osteosarcoma. Treatment with BEMPEG delayed tumor growth and increased overall survival of mice with K7M2-WT osteosarcoma pulmonary metastases. BEMPEG also inhibited primary tumor growth and metastatic relapse in lungs and bone in the K7M3 orthotopic osteosarcoma mouse model. In addition, it enhanced therapeutic activity of anti-CTLA-4 and anti-PD-1 checkpoint blockade in the DLM8 subcutaneous murine osteosarcoma model. Finally, BEMPEG strongly increased accumulation of intratumoral effector T cells and natural killer cells, but not T-regulatory cells, resulting in improved effector:inhibitory cell ratios. Collectively, these data in multiple murine models of osteosarcoma provide a path toward clinical evaluation of BEMPEG-based regimens in human osteosarcoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Óseas/tratamiento farmacológico , Modelos Animales de Enfermedad , Interleucina-2/análogos & derivados , Osteosarcoma/tratamiento farmacológico , Polietilenglicoles/farmacología , Animales , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Línea Celular Tumoral , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Interleucina-2/administración & dosificación , Interleucina-2/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Osteosarcoma/inmunología , Osteosarcoma/patología , Polietilenglicoles/administración & dosificación , Análisis de Supervivencia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
3.
Ann Biomed Eng ; 49(2): 548-559, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32761557

RESUMEN

Ovarian cancer is the most lethal malignancy affecting the female reproductive system. Identification and removal of all ovarian intraperitoneal tumor deposits during the intraoperative surgery is important towards preventing cancer recurrence and ultimately improving patient survival. Herein, we investigate the effectiveness of virus mimicking nanoparticles, derived from genome-depleted plant-infecting brome mosaic virus, and doped with near infrared (NIR) brominated cyanine dye BrCy106-NHS, for targeted NIR fluorescence imaging of intraperitoneal ovarian tumors. We refer to these nanoparticles as optical viral ghosts (OVGs). We functionalized the OVGs with antibodies against HER2 receptor, a biomarker over-expressed in ovarian cancers. We injected functionalized OVGs, non-functionalized OVGs, and non-encapsulated BrCy106-NHS intravenously in mice implanted with ovarian intraperitoneal tumors. Tumors were extracted at 2, 6, and 24 h post-injection, and quantitatively analyzed using NIR fluorescence imaging. Fluorescence emission from tumors associated with the injection of the functionalized OVGs continued to increase between 2 and 24 h post-injection. At 24 h timepoint, the average spectrally-integrated fluorescence emission from homogenized tumors containing functionalized-OVGs was about 3.5 and 19.5 times higher than those containing non-functionalized OVGs or non-encapsulated BrCy106-NHS, respectively. Similarly, by using the functionalized-OVGs, the imaging signal-to-noise ratio at 24 h timepoint was enhanced by approximately threefold and sevenfold as compared to non-functionalized OVGs and the non-encapsulated dye, respectively. These functionalized virus-mimicking NIR nano-constructs could potentially be used for intraoperative visualization of ovarian tumors implants.


Asunto(s)
Bromovirus , Colorantes Fluorescentes/administración & dosificación , Nanopartículas/administración & dosificación , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Peritoneales/diagnóstico por imagen , Receptor ErbB-2 , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones Desnudos , Imagen Óptica/métodos , Trasplante Heterólogo
4.
Clin Cancer Res ; 26(18): 4933-4946, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32576626

RESUMEN

PURPOSE: Advances in prostate cancer lag behind other tumor types partly due to the paucity of models reflecting key milestones in prostate cancer progression. Therefore, we develop clinically relevant prostate cancer models. EXPERIMENTAL DESIGN: Since 1996, we have generated clinically annotated patient-derived xenografts (PDXs; the MDA PCa PDX series) linked to specific phenotypes reflecting all aspects of clinical prostate cancer. RESULTS: We studied two cell line-derived xenografts and the first 80 PDXs derived from 47 human prostate cancer donors. Of these, 47 PDXs derived from 22 donors are working models and can be expanded either as cell lines (MDA PCa 2a and 2b) or PDXs. The histopathologic, genomic, and molecular characteristics (androgen receptor, ERG, and PTEN loss) maintain fidelity with the human tumor and correlate with published findings. PDX growth response to mouse castration and targeted therapy illustrate their clinical utility. Comparative genomic hybridization and sequencing show significant differences in oncogenic pathways in pairs of PDXs derived from different areas of the same tumor. We also identified a recurrent focal deletion in an area that includes the speckle-type POZ protein-like (SPOPL) gene in PDXs derived from seven human donors of 28 studied (25%). SPOPL is a SPOP paralog, and SPOP mutations define a molecular subclass of prostate cancer. SPOPL deletions are found in 7% of The Cancer Genome Atlas prostate cancers, which suggests that our cohort is a reliable platform for targeted drug development. CONCLUSIONS: The MDA PCa PDX series is a dynamic resource that captures the molecular landscape of prostate cancers progressing under novel treatments and enables optimization of prostate cancer-specific, marker-driven therapy.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor/genética , Medicina de Precisión/métodos , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Humanos , Masculino , Ratones , Cultivo Primario de Células , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Eliminación de Secuencia , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Biotechniques ; 69(1): 395-403, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32363906

RESUMEN

Methods for imaging orthotopic prostate tumors within the prostate or small tumors with extension outside the prostate are needed to more closely model human prostate tumors, which are most commonly located within the gland or may extend just through the gland. By comparing MR sequences, we found that the T2-based Dixon 'water only' sequence best visualized tumors within the prostate of mouse models in both young and old mice and that tumor weight derived from this sequence correlated highly with ex vivo tumor weight (r2 = 0.98, p < 0.001, n = 12). This should aid tumor detection, margin delineation and evaluation of tumor burden to enable studies including, but not limited to, evaluating the natural history of the disease, the mechanisms of action and the efficacy of therapeutic interventions.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Próstata/patología , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Desnudos , Células PC-3 , Carga Tumoral/fisiología
6.
Mol Pharm ; 16(2): 808-815, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30608713

RESUMEN

Metformin is the most widely prescribed drug for type 2 diabetes. Chemically, metformin is a hydrophilic base that functions as an organic cation, suggesting that it may have the capacity to inhibit the tubular reabsorption of peptide radiotracers. The purpose of this study was to investigate whether metformin could reduce renal uptake of peptidyl radiotracers and serve as a radioprotective agent for peptide receptor radionuclide therapy (PRRT). METHODS: We used two radiolabeled peptides: a 68Ga-labeled cyclic (TNYL-RAW) peptide (68Ga-NOTA-c(TNYL-RAW) (NOTA: 1,4,7 triazacyclononane-1,4,7-trisacetic acid) targeting EphB4 receptors and an 111In- or 64Cu-labeled octreotide (111In/64Cu-DOTA-octreotide) (DOTA: 1,4,7,10 triazacyclododecane-1,4,7,10-tetraacetic acid) targeting somatostatin receptors. Each radiotracer was injected intravenously into normal Swiss mice or tumor-bearing nude mice in the presence or absence of metformin administered intravenously or orally. Micropositron emission tomography or microsingle-photon emission computed tomography images were acquired at different times after radiotracer injection, and biodistribution studies were performed at the end of the imaging session. To assess the radioprotective effect of metformin on the kidneys, normal Swiss mice received two doses of 111In-DOTA-octreotidein the presence or absence of metformin, and renal function was analyzed via blood chemistry and histology. RESULTS: Intravenous injection of metformin with 68Ga-NOTA-c(TNYL-RAW) or 111In-DOTA-octreotide reduced the renal uptake of the radiotracer by 60% and 35%, respectively, compared to uptake without metformin. These reductions were accompanied by greater uptake in the tumors for both radiolabeled peptides. Moreover, the renal uptake of 111In-DOTA-octreotide was significantly reduced when metformin was administered via oral gavage. Significantly more radioactivity was recovered in the urine collected over a period of 24 h after intravenous injection of 64Cu-DOTA-octreotide in mice that received oral metformin than in mice that received vehicle. Finally, coadministration of 111In-DOTA-octreotide with metformin mitigated radio-nephrotoxicity. CONCLUSION: Metformin inhibits kidney uptake of peptidyl radiotracers, protecting the kidney from nephrotoxicity. Further studies are needed to elucidate the mechanisms of these finding and to optimize mitigation of radiation-induced damage to kidney in PRRT.


Asunto(s)
Riñón/metabolismo , Metformina/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Línea Celular Tumoral , Radioisótopos de Cobre/metabolismo , Femenino , Humanos , Inyecciones Intravenosas , Riñón/efectos de los fármacos , Metformina/administración & dosificación , Ratones , Ratones Desnudos , Octreótido/metabolismo , Tomografía de Emisión de Positrones , Receptores de Péptidos/metabolismo , Tomografía Computarizada de Emisión de Fotón Único
7.
ACS Nano ; 12(8): 8214-8223, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30088917

RESUMEN

Engineering a compact, near-infrared plasmonic nanostructure with integrated image-enhancing agents for combined imaging and therapy is an important nanomedical challenge. Recently, we showed that Au@SiO2@Au nanomatryoshkas (NM) are a highly promising nanostructure for hosting either T1 MRI or fluorescent contrast agents with a photothermal therapeutic response in a compact geometry. Here, we show that a near-infrared-resonant NM can provide simultaneous contrast enhancement for both T1 magnetic resonance imaging (MRI) and fluorescence optical imaging (FOI) by encapsulating both types of contrast agents in the internal silica layer between the Au core and shell. We also show that this method of T1 enhancement is even more effective for Fe(III), a potentially safer contrast agent compared to Gd(III). Fe-NM-based contrast agents are found to have relaxivities 2× greater than those found in the widely used gadolinium chelate, Gd(III) DOTA, providing a practical alternative that would eliminate Gd(III) patient exposure entirely. This dual-modality nanostructure can enable not only tissue visualization with MRI but also fluorescence-based nanoparticle tracking for quantifying nanoparticle distributions in vivo, in addition to a near-infrared photothermal therapeutic response.


Asunto(s)
Medios de Contraste/química , Fluorescencia , Imagen por Resonancia Magnética , Nanopartículas del Metal/química , Animales , Gadolinio/química , Oro/química , Hierro/química , Manganeso/química , Ratones , Imagen Óptica , Fototerapia , Dióxido de Silicio/química
8.
Radiology ; 285(3): 830-838, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28707963

RESUMEN

Purpose To assess in a mouse model whether early or late components of glucose metabolism, exemplified by fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography (PET) and hyperpolarized carbon 13 (13C)-pyruvate magnetic resonance (MR) spectroscopy, can serve as indicators of response in ovarian cancer to multityrosine kinase inhibitor pazopanib. Materials and Methods In this Animal Care and Use Committee approved study, 17 days after the injection of 2 × 106 human ovarian SKOV3 tumors cells into 14 female nude mice, treatment with vehicle or pazopanib (2.5 mg per mouse peroral every other day) was initiated. Longitudinal T2-weighted MR imaging, dynamic MR spectroscopy of hyperpolarized pyruvate, and 18F-FDG PET/computed tomographic (CT) imaging were performed before treatment, 2 days after treatment, and 2 weeks after treatment. Results Pazopanib inhibited ovarian tumor growth compared with control (0.054 g ± 0.041 vs 0.223 g ± 0.112, respectively; six mice were treated with pazopanib and seven were control mice; P < .05). Significantly higher pyruvate-to-lactate conversion (lactate/pyruvate + lactate ratio) was found 2 days after treatment with pazopanib than before treatment (0.46 ± 0.07 vs 0.31 ± 0.14, respectively; P < .05; six tumors after treatment, seven tumors before treatment). This was not observed with the control group or with 18F-FDG PET/CT imaging. Conclusion The findings suggest that hyperpolarized 13C-pyruvate MR spectroscopy may serve as an early indicator of response to tyrosine kinase (angiogenesis) inhibitors such as pazopanib in ovarian cancer even when 18F-FDG PET/CT does not indicate a response. © RSNA, 2017 Online supplemental material is available for this article.


Asunto(s)
Espectroscopía de Resonancia Magnética con Carbono-13/métodos , Monitoreo de Drogas/métodos , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/tratamiento farmacológico , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Pirimidinas/administración & dosificación , Sulfonamidas/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Femenino , Fluorodesoxiglucosa F18 , Humanos , Indazoles , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Desnudos , Imagen Molecular/métodos , Imagen Multimodal/métodos , Evaluación de Resultado en la Atención de Salud/métodos , Neoplasias Ováricas/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Radiofármacos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Resultado del Tratamiento
9.
EJNMMI Res ; 6(1): 25, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26983635

RESUMEN

BACKGROUND: Gene therapy has been hampered by low expression upon in vivo delivery. Using a somatostatin receptor type 2 (SSTR2)-based reporter, we assessed whether angiotensin II (AII) can improve gene expression by adenovirus upon intra-arterial (IA) delivery in a large animal model. METHODS: A SSTR2-based reporter that can be imaged by a clinically approved radiopharmaceutical was used to assess gene expression. Eight rabbits bearing VX2 tumors in each thigh were randomly injected IA with adenovirus containing a human SSTR2 (Ad-CMV-HA-SSTR2) gene chimera ± AII or control adenovirus containing green fluorescent protein (Ad-CMV-GFP). Three days later, (111)In-octreotide was given IV after computed tomography (CT) imaging using a clinical CT scanner and intravenous contrast. Tumor uptake of (111)In-octreotide was evaluated the next day using a clinical gamma camera. Gene expression was normalized to tumor weight and morphology from CT to obtain in vivo biodistribution. RESULTS: SSTR2-based expression was readily visualized. VX2 tumors infected with Ad-CMV-HA-SSTR2 upon intra-arterial delivery with AII had greater in vivo biodistribution, thus greater gene expression, than those without AII (p < 0.01, n = 6). VX2 tumors infected with Ad-CMV-HA-SSTR2 upon IA delivery had greater biodistribution, thus greater gene expression, than those with the negative control Ad-CMV-GFP (p < 0.02). Similarly, VX2 tumors infected with Ad-CMV-HA-SSTR2 upon IA delivery with AII had greater biodistribution, thus greater gene expression, than those with the negative control Ad-CMV-GFP (p < 0.01). CONCLUSIONS: Angiotensin II improves in vivo gene expression by adenovirus upon intra-arterial delivery and thus may improve gene therapy efficacy. In vivo SSTR2-based reporter imaging can be used to compare methodologies for improving gene expression.

10.
Oncotarget ; 6(36): 38816-26, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26452035

RESUMEN

The purpose of this study was to identify the role COX-2 plays in K-ras-induced lung carcinogenesis. We crossed COX-2-homozygous knockout mice with K-rasLA1 (G12D) expressing mice to obtain COX-2-deficient mice with K-ras expression (K-ras/COX-2(-/-) mice) and COX-2 wild type mice with K-ras expression (K-ras mice). At 3.5 months of age, the K-ras/COX-2(-/-) mice had significantly fewer lung adenocarcinomas and substantially smaller tumors than K-ras mice. K-ras/COX-2(-/-) mice also had significantly fewer bronchioalveolar hyperplasias than K-ras mice. Compared with lung tumors from K-Ras mice, the levels of prostaglandin E2 (PGE2) were significantly lower, whereas levels of the PGE2 metabolite 13,14-dihydro-15-keto-PGE2 were significantly higher, in lung tumors from K-ras/COX-2(-/-) mice. In addition, K-ras/COX-2(-/-) mice had strikingly lower rates of tumor cell proliferation and expressed less MEK and p-Erk1/2 protein than K-ras mice did. In line with this, knocking down COX-2 in mutant K-ras non-small cell lung cancer A549 cells reduced colony formation, PGE2 synthesis and ERK phosphorylation compared to that of vector control cells. Taken together, these findings suggest that COX-2 deletion contributes to the repression of K-ras-induced lung tumorigenesis by reducing tumor cell proliferation, decreasing the production of PGE2, and increasing the production of 13,14-dihydro-15-keto-PGE2, possibly via the MAPK pathway. Thus, COX-2 is likely important in lung tumorigenesis, and COX-2 and its product, PGE2, are potential targets for lung cancer prevention.


Asunto(s)
Adenocarcinoma/enzimología , Carcinogénesis/metabolismo , Ciclooxigenasa 2/metabolismo , Neoplasias Pulmonares/enzimología , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/genética , Dinoprostona/biosíntesis , Femenino , Técnicas de Inactivación de Genes , Genes ras , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal
11.
PLoS One ; 10(7): e0130168, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26208254

RESUMEN

BACKGROUND: Dynamic contrast-enhanced MRI (DCE-MRI) biomarkers have proven utility in tumors in evaluating microvascular perfusion and permeability, but it is unclear whether measurements made in different centers are comparable due to methodological differences. PURPOSE: To evaluate how commonly utilized analytical methods for DCE-MRI biomarkers affect both the absolute parameter values and repeatability. MATERIALS AND METHODS: DCE-MRI was performed on three consecutive days in twelve rats bearing C6 xenografts. Endothelial transfer constant (Ktrans), extracellular extravascular space volume fraction (ve), and contrast agent reflux rate constant (kep) measures were computed using: 2-parameter ("Tofts" or "standard Kety") vs. 3-parameter ("General Kinetic" or "extended Kety") compartmental models (including blood plasma volume fraction (vp) with 3-parameter models); individual- vs. population-based vascular input functions (VIFs); and pixel-by-pixel vs. whole tumor-ROI. Variability was evaluated by within-subject coefficient of variation (wCV) and variance components analyses. RESULTS: DCE-MRI absolute parameter values and wCVs varied widely by analytical method. Absolute parameter values ranged, as follows, median Ktrans, 0.09-0.18 min-1; kep, 0.51-0.92 min-1; ve, 0.17-0.23; and vp, 0.02-0.04. wCVs also varied widely by analytical method, as follows: mean Ktrans, 32.9-61.9%; kep, 11.6-41.9%; ve, 16.1-54.9%; and vp, 53.9-77.2%. Ktrans and kep values were lower with 3- than 2-parameter modeling (p<0.0001); kep and vp were lower with pixel- than whole-ROI analyses (p<0.0006). wCVs were significantly smaller for ve, and larger for kep, with individual- than population-based VIFs. CONCLUSIONS: DCE-MRI parameter values and repeatability can vary widely by analytical methodology. Absolute values of DCE-MRI biomarkers are unlikely to be comparable between different studies unless analyses are carefully standardized.


Asunto(s)
Algoritmos , Biomarcadores/análisis , Aumento de la Imagen/métodos , Imagen por Resonancia Magnética/métodos , Animales , Línea Celular Tumoral , Medios de Contraste , Glioma/diagnóstico , Glioma/diagnóstico por imagen , Masculino , Neoplasias Experimentales/diagnóstico , Neoplasias Experimentales/diagnóstico por imagen , Radiografía , Ratas , Ratas Desnudas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
12.
PLoS One ; 10(6): e0131095, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26098849

RESUMEN

PURPOSE: To assess whether T1 relaxation time of tumors may be used to assess response to bevacizumab anti-angiogenic therapy. PROCEDURES: 12 female nude mice bearing subcutaneous SKOV3ip1-LC ovarian tumors were administered bevacizumab (6.25ug/g, n=6) or PBS (control, n=6) therapy twice a week for two weeks. T1 maps of tumors were generated before, two days, and 2 weeks after initiating therapy. Tumor weight was assessed by MR and at necropsy. Histology for microvessel density, proliferation, and apoptosis was performed. RESULTS: Bevacizumab treatment resulted in tumor growth inhibition (p<0.04, n=6), confirming therapeutic efficacy. Tumor T1 relaxation times increased in bevacizumab treated mice 2 days and 2 weeks after initiating therapy (p<.05, n=6). Microvessel density decreased 59% and cell proliferation (Ki67+) decreased 50% in the bevacizumab treatment group (p<.001, n=6), but not apoptosis. CONCLUSIONS: Findings suggest that increased tumor T1 relaxation time is associated with response to bevacizumab therapy in ovarian cancer model and might serve as an early indicator of response.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Bevacizumab/uso terapéutico , Neoplasias Ováricas/terapia , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Imagen por Resonancia Magnética , Ratones , Ratones Desnudos , Microvasos/patología , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Invest New Drugs ; 33(2): 271-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25476893

RESUMEN

Introduction Oleandrin, a cardiac glycoside, exerts strong anti-proliferative activity against various human malignancies in in vitro cells. Here, we report the antitumor efficacy of PBI-05204, a supercritical C02 extract of Nerium oleander containing oleandrin, in a human pancreatic cancer Panc-1 orthotopic model. Results While all the control mice exhibited tumors by the end of treatment, only 2 of 8 mice (25%) treated for 6 weeks with PBI-05204 (40 mg/kg) showed dissectible tumor at the end of the treatment period. The average tumor weight (222.9 ± 116.9 mg) in mice treated with PBI-05204 (20 mg/kg) was significantly reduced from that in controls (920.0 ± 430.0 mg) (p < 0.05). Histopathologic examination of serial sections from each pancreas with no dissectible tumor in the PBI-05204 (40 mg/kg) treated group showed that the pancreatic tissues of 5/6 mice were normal while the remaining mouse had a tumor the largest diameter of which was less than 2.3 mm. In contrast, while gemcitabine alone did not significantly reduce tumor growth, PBI-05204 markedly enhanced the antitumor efficacy of gemcitabine in this particular model. Ki-67 staining was reduced in pancreatic tumors from mice treated with PBI-05204 (20 mg/kg) compared to that of control, suggesting that PBI-05204 inhibited the proliferation of the Panc-1 tumor cells. PBI-05204 suppressed expression of pAkt, pS6, and p4EPB1 in a concentration-dependent manner in both Panc-1 tumor tissues and human pancreatic cancer cell lines, implying that this novel botanical drug exerts its potent antitumor activity, at least in part, through down-regulation of PI3k/Akt and mTOR pathways.


Asunto(s)
Glicósidos Cardíacos/farmacología , Fosfatidilinositol 3-Quinasa Clase Ib/biosíntesis , Nerium , Neoplasias Pancreáticas/tratamiento farmacológico , Extractos Vegetales/farmacología , Serina-Treonina Quinasas TOR/biosíntesis , Animales , Ciclo Celular , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Regulación hacia Abajo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Gemcitabina
14.
Sci Transl Med ; 6(252): 252ra122, 2014 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-25186177

RESUMEN

Bone is the most common site of prostate cancer (PCa) progression to a therapy-resistant, lethal phenotype. We found that blockade of fibroblast growth factor receptors (FGFRs) with the receptor tyrosine kinase inhibitor dovitinib has clinical activity in a subset of men with castration-resistant PCa and bone metastases. Our integrated analyses suggest that FGF signaling mediates a positive feedback loop between PCa cells and bone cells and that blockade of FGFR1 in osteoblasts partially mediates the antitumor activity of dovitinib by improving bone quality and by blocking PCa cell-bone cell interaction. These findings account for clinical observations such as reductions in lesion size and intensity on bone scans, lymph node size, and tumor-specific symptoms without proportional declines in serum prostate-specific antigen concentration. Our findings suggest that targeting FGFR has therapeutic activity in advanced PCa and provide direction for the development of therapies with FGFR inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Bencimidazoles/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Neoplasias de la Próstata/patología , Quinolonas/uso terapéutico , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Bencimidazoles/farmacología , Neoplasias Óseas/patología , Huesos/efectos de los fármacos , Huesos/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Quinolonas/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Nat Commun ; 4: 2427, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24018975

RESUMEN

The miR-200 family is well known to inhibit the epithelial-mesenchymal transition, suggesting it may therapeutically inhibit metastatic biology. However, conflicting reports regarding the role of miR-200 in suppressing or promoting metastasis in different cancer types have left unanswered questions. Here we demonstrate a difference in clinical outcome based on miR-200's role in blocking tumour angiogenesis. We demonstrate that miR-200 inhibits angiogenesis through direct and indirect mechanisms by targeting interleukin-8 and CXCL1 secreted by the tumour endothelial and cancer cells. Using several experimental models, we demonstrate the therapeutic potential of miR-200 delivery in ovarian, lung, renal and basal-like breast cancers by inhibiting angiogenesis. Delivery of miR-200 members into the tumour endothelium resulted in marked reductions in metastasis and angiogenesis, and induced vascular normalization. The role of miR-200 in blocking cancer angiogenesis in a cancer-dependent context defines its utility as a potential therapeutic agent.


Asunto(s)
MicroARNs/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/genética , Neovascularización Patológica/genética , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Movimiento Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Neoplasias Pulmonares/secundario , MicroARNs/genética , Modelos Biológicos , Nanopartículas/administración & dosificación , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Oligonucleótidos/farmacología , Oligonucleótidos/uso terapéutico , Pericitos/efectos de los fármacos , Pericitos/patología , Resultado del Tratamiento
16.
PLoS One ; 8(6): e62371, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23762226

RESUMEN

PURPOSE: To evaluate the importance of morphology in quantifying expression after in vivo gene transfer and to compare gene expression after intra-arterial (IA) and intra-tumoral (IT) delivery of adenovirus expressing a SSTR2-based reporter gene in a large animal tumor model. MATERIALS AND METHODS: Tumor directed IA or IT delivery of adenovirus containing a human somatostatin receptor type 2A (Ad-CMV-HA-SSTR2A) gene chimera or control adenovirus (Ad-CMV-GFP) was performed in VX2 tumors growing in both rabbit thighs. Three days later, ¹¹¹In-octreotide was administered intravenously after CT imaging using a clinical scanner. ¹¹¹In-octreotide uptake in tumors was evaluated the following day using a clinical gamma-camera. Gene expression was normalized to tumor weight with and without necrosis. This procedure was repeated on nine additional rabbits to investigate longitudinal gene expression both 5 days and 2 weeks after adenovirus delivery. CT images were used to evaluate tumor morphology and excised tissue samples were analyzed to determine ¹¹¹In-octreotide biodistribution ex vivo. RESULTS: VX2 tumors infected with Ad-CMV-HA-SSTR2 had greater ¹¹¹In-octreotide uptake than with control virus (P<0.05). Intra-arterial and intra-tumoral routes resulted in similar levels of gene expression. Longitudinally, expression appeared to wane at 2 weeks versus 5 days after delivery. Areas of necrosis did not demonstrate significant uptake ex vivo. Morphology identified areas of necrosis on contrast enhanced CT and upon excluding necrosis, in vivo biodistribution analysis resulted in greater percent injected dose per gram (P<0.01) and corresponded better with ex vivo biodistribution(r = 0.72, P<0.01, Coefficient of the x-variable = .72) at 2 weeks than without excluding necrosis (P<0.01). CONCLUSION: Tumor specificity and high transgene expression can be achieved in tumors via both tumor directed intra-arterial and intra-tumoral delivery in a large animal tumor model. Using clinical machines, morphologic imaging contributes to functional imaging for quantifying SSTR2-based reporter expression in vivo.


Asunto(s)
Adenoviridae/genética , Carcinoma Adenoescamoso/patología , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/administración & dosificación , Octreótido/análogos & derivados , Receptores de Somatostatina/genética , Animales , Western Blotting , Carcinoma Adenoescamoso/genética , Carcinoma Adenoescamoso/terapia , Vías de Administración de Medicamentos , Cámaras gamma , Genes Reporteros/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Procesamiento de Imagen Asistido por Computador , Inyecciones Intraarteriales , Inyecciones Intralesiones , Necrosis , Octreótido/farmacocinética , Conejos , Radiofármacos/farmacocinética , Receptores de Somatostatina/metabolismo , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único , Transgenes/fisiología , Carga Tumoral , Células Tumorales Cultivadas
17.
Bone ; 50(3): 695-703, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22173053

RESUMEN

Transforming growth factor beta 1 (TGF-ß1) has been implicated in the pathogenesis of prostate cancer (PCa) bone metastasis. In this study, we tested the antitumor efficacy of a selective TGF-ß receptor I kinase inhibitor, LY2109761, in preclinical models. The effect of LY2109761 on the growth of MDA PCa 2b and PC-3 human PCa cells and primary mouse osteoblasts (PMOs) was assessed in vitro by measuring radiolabeled thymidine incorporation into DNA. In vivo, the right femurs of male SCID mice were injected with PCa cells. We monitored the tumor burden in control- and LY2109761-treated mice with MRI analysis and the PCa-induced bone response with X-ray and micro-CT analyses. Histologic changes in bone were studied by performing bone histomorphometric evaluations. PCa cells and PMOs expressed TGF-ß receptor I. TGF-ß1 induced pathway activation (as assessed by induced expression of p-Smad2) and inhibited cell growth in PC-3 cells and PMOs but not in MDA PCa 2b cells. LY2109761 had no effect on PCa cells but induced PMO proliferation in vitro. As expected, LY2109761 reversed the TGF-ß1-induced pathway activation and growth inhibition in PC-3 cells and PMOs. In vivo, LY2109761 treatment for 6weeks resulted in increased volume in normal bone and increased osteoblast and osteoclast parameters. In addition, LY2109761 treatment significantly inhibited the growth of MDA PCa 2b and PC-3 in the bone of SCID mice (p<0.05); moreover, it resulted in significantly less bone loss and change in osteoclast-associated parameters in the PC-3 tumor-bearing bones than in the untreated mice. In summary, we report for the first time that targeting TGF-ß receptors with LY2109761 can control PCa bone growth while increasing the mass of normal bone. This increased bone mass in nontumorous bone may be a desirable side effect of LY2109761 treatment for men with osteopenia or osteoporosis secondary to androgen-ablation therapy, reinforcing the benefit of effectively controlling PCa growth in bone. Thus, targeting TGF-ß receptor I is a valuable intervention in men with advanced PCa.


Asunto(s)
Antineoplásicos/farmacología , Huesos/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirroles/farmacología , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Animales , Neoplasias Óseas/secundario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fémur/efectos de los fármacos , Fémur/metabolismo , Fémur/patología , Humanos , Masculino , Ratones , Osteoblastos/metabolismo , Osteoblastos/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Radiology ; 260(3): 718-26, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21633050

RESUMEN

PURPOSE: To investigate the effects of increasing doses of angiotensin II on hepatic hemodynamics in the normal rabbit liver and in hepatic VX2 tumors by using dynamic contrast material-enhanced perfusion computed tomography (CT). MATERIALS AND METHODS: This study was approved by the institutional animal care and use committee. Solitary hepatic VX2 tumors were implanted into 12 rabbits. In each animal, perfusion CT of the liver was performed before (at baseline) and after hepatic arterial infusion of varying doses (0.1-50.0 µg/mL) of angiotensin II. Images were acquired continuously for 80 seconds after the start of the intravenous contrast material administration. Blood flow (BF), blood volume (BV), mean transit time (MTT), and capillary permeability-surface area product were calculated for the tumor and the adjacent and distant normal liver tissue. Generalized linear mixed models were used to estimate the effects of angiotensin II dose on outcome measures. RESULTS: Angiotensin II infusion increased contrast enhancement of the tumor and distal liver vessels. Tumor BF increased in a dose-dependent manner after administration of 0.5-25.0 µg/mL angiotensin II, but only the 2.5 µg/mL dose induced a significant increase in tumor BF compared with BF in the adjacent (68.0 vs 26.3 mL/min/100 g, P < .0001) and distant (68.0 vs 28.3 mL/min/100 g, P = .02) normal liver tissue. Tumor BV varied with angiotensin II dose but was greater than the BV of the adjacent and distant liver tissue at only the 2.5 µg/mL (4.8 vs 3.5 mL/100 g for adjacent liver [P < .0001], 4.8 vs 3.3 mL/100 g for distant liver [P = .0006]) and 10.0 µg/mL (4.9 vs 4.4 mL/100 g for adjacent liver [P = .007], 4.9 vs 4.3 mL/100 g for distant liver [P = .04]) doses. Tumor MTT was significantly shorter than the adjacent liver tissue MTT at angiotensin II doses of 2.5 µg/mL (9.7 vs 15.8 sec, P = .001) and 10.0 µg/mL (5.1 vs 13.2 sec, P = .007) and significantly shorter than the distant liver tissue MTT at 2.5 µg/mL only (9.7 vs 15.3 sec, P = .0006). The capillary permeability-surface area product for the tumor was higher than that for the adjacent liver tissue at the 2.5 µg/mL angiotensin II dose only (11.5 vs 8.1 mL/min/100 g, P = .01). CONCLUSION: Perfusion CT enables a mechanistic understanding of angiotensin II infusion in the liver and derivation of the optimal effective dose. The 2.5 µg/mL angiotensin II dose increases perfusion in hepatic VX2 tumors versus that in adjacent and distant normal liver tissue primarily by constricting normal distal liver vessels and in turn increasing tumor BF and BV.


Asunto(s)
Angiotensina II/administración & dosificación , Circulación Hepática , Neoplasias Hepáticas Experimentales/diagnóstico por imagen , Neoplasias Hepáticas Experimentales/fisiopatología , Imagen de Perfusión/métodos , Tomografía Computarizada por Rayos X/métodos , Animales , Velocidad del Flujo Sanguíneo , Medios de Contraste/administración & dosificación , Aumento de la Imagen/métodos , Infusiones Intralesiones , Conejos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Vasoconstrictores/administración & dosificación
19.
Cancer Cell ; 18(2): 185-97, 2010 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-20708159

RESUMEN

Although VEGF-targeted therapies are showing promise, new angiogenesis targets are needed to make additional gains. Here, we show that increased Zeste homolog 2 (EZH2) expression in either tumor cells or in tumor vasculature is predictive of poor clinical outcome. The increase in endothelial EZH2 is a direct result of VEGF stimulation by a paracrine circuit that promotes angiogenesis by methylating and silencing vasohibin1 (vash1). Ezh2 silencing in the tumor-associated endothelial cells inhibited angiogenesis mediated by reactivation of VASH1, and reduced ovarian cancer growth, which is further enhanced in combination with ezh2 silencing in tumor cells. Collectively, these data support the potential for targeting ezh2 as an important therapeutic approach.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/fisiología , Neovascularización Patológica/fisiopatología , Neoplasias Ováricas/irrigación sanguínea , Animales , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN , Cartilla de ADN , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/genética , Inmunohistoquímica , Ratones , Ratones Desnudos , Microscopía Fluorescente , Neoplasias Ováricas/patología , Complejo Represivo Polycomb 2
20.
Transl Oncol ; 2(2): 96-106, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19412425

RESUMEN

Small animal computed tomography (CT) has poor intrinsic soft tissue contrast, limiting evaluation of intra-abdominal structures. Using standard intravascular-extracellular intravenous contrast (IE-IV) alone is theoretically limited by long acquisition times of traditional small animal scanners that may result in equilibration. We assessed whether a negative contrast strategy of enhancing normal tissue surrounding tumor, instead of the tumor itself, can visualize and quantify intraperitoneal (IP) cancer in a mouse model. Two and a half weeks after IP injection of Hey A8 cells, four groups of three animals each were administered serial dilutions of IV Fenestra LC (RES-IV), oral Gastroview, and IP Optiray 320. Another group of three animals was administered IV Optiray 320 (IE-IV), oral Gastroview, and IP Optiray 320 in successive combinations. Both groups were imaged by CT. Tumor and organ Hounsfield units were measured, and visualization was assessed. With increasing contrast amount, the Hounsfield unit of organs generally increased, whereas that of tumor remained essentially stable. The visualization of abdominal organs and tumor also generally increased with increasing contrast amount. Visualization of tumor and its margins adjacent to liver, spleen, and stomach was significantly better on administering RES-IV. However, for tumor adjacent to bladder, both IE-IV and RES-IV were equivalent. In vivo CT-derived tumor weights correlated highly with ex vivo tumor weights (r = 0.96, P < .0001, n = 15). Thus, CT using negative contrast enhancement strategy allows visualization and quantification of IP tumors. Such a strategy will also enable anatomic localization of functional signal for combination/molecular imaging.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...